REVIEW The advantages of nanobodies and the prospects these offer in cancer treatment Peter van Doorn Universiteit Utrecht, Thesis for the master Molecular and Cellular Life Sciences Abstract: VHH fragments, also called nanobodies, derived from heavy-chain only antibodies (HCAb) originally found in camels, are the so far smallest known antibody fragments that are still capable of antigen binding with high specificity. These nanobodies have properties that make them superior in many ways compared to conventional antibodies. They are therefore being used in an increasing number of varying applications ranging from uses in consumer products because of their low costs of manufacturing to the inhibition of enzymes because of their unique binding capabilities and small size. In therapeutics, however, nanobodies receive special attention in the treatment and diagnosis of cancer. Properties like better tissue penetration make them suitable for the treatment of solid tumors and their monomeric nature for instance opens up other strategies in cancer therapeutics. In this review an overview is presented of several advantages that nanobodies have together with some of the applications that make use of these. The emphasis of the possible uses of nanobodies, however, is on their use in cancer therapy. The mentioned advantages of nanobodies are described in relation to possible new and improved applications they offer in regard to cancer treatment. Introduction Since the approval in therapeutics of the first antibody in 1986 antibodies have been an important focus in drug development. This first drug started with the conventional Immunoglobulin G (IgG), the most abundant type of antibody. IgG are tetramers of 150 kDa consisting of two identical heavy chains that are each 50 kDa and 2 identical light chains of 25 kDa. These chains are linked to each other by disulfide bonds [1]. For antigen binding these immunoglobulins require a variable domain at the N-terminal end of the light chain and one at the heavy chain [1]. High production costs and other downsides like their large size and relative instability however led research to smaller antibody derived fragments. The first ones for instance were the Fab fragments (~55 kDa) that consist only of the light and heavy chain, without the 2 constant domains CH2 and CH3 (Figure 1a). Later Fv fragments consisting only of the variable heavy (VH : 13.6 kDa) and variable light (VL : 12.4 kDa) chain were made (Figure 1a) [2]. These Fv fragments however were found to dissociate upon dilution and had to be linked together by a single-chain (sc), resulting in the scFv fragments (~28 kDa) (Figure 1a)[3]. Antibodies consisting of only the VH domain Figure 1. (A) Schematic representation of a conventional antibody and its antibody derived fragments. (B) Schematic representation of a heavy-chain only antibody and the VHH derived from these. HCAb (Heavy-chain only antibody), CH (constant heavy domain), CL (constant light domain), VH (variable heavy domain), VL (variable light domain), VHH (variable heavy domain from HCAb). 1 (~12-15 kDa) from normal IgG have also been tested [4]. And although these even smaller VH domains do retain some antigen specificity (even light chain only fragments were found to retain activity [5]) the removal of the VL domain from a Fv fragment exposed a too large hydrophobic surface. This resulted in the VH molecules to aggregate and consequently their affinity to drop even further than that of scFv which already had a lower affinity than conventional IgG [6]. Fortunately many of the problems the antibody derived fragments had were solved when it was found that designing smaller antibodies was not something that had to be done in the lab since it was already occurring in nature. ly removed by the light chain. BiP however retains the heavy chain by a stable interaction with the CH1 domain [11]. Differences between VH and VHH Because VH from mice or humans and VHH from Camelidae are all originating from vertebrates they show a not unsuspected high degree of similarity between their genes. Why it then is that these VHH are functional were VH fragments are not, or at least in a much smaller degree, lies in certain structural adaptations that VHH underwent. VHH are different from normal VH because of the substitution of a number of amino acids that make the before mentioned hydrophobic surface of the VH more hydrophilic [12]. The positive effect of these mutations in HCAbs was corroborated by Davies & Riechmann when they ‘camelised’ human antibody fragments by mutating 3 of these amino acids and showed a significantly The structure of HCAb Although the structure of antibodies is highly conserved among mammals several variations have been found that lack the VL chain and only have the VH chain. These antibodies have for instance been seen in the heavy chain disease [7] and in cartilaginous fish like sharks [8] wherein naturally occuring heavy-chain only antibodies (HCAb) exist. The third type of HCAb is the one receiving the most attention and was first found by Hamers-Casterman et al. in camels [9]. The VH fragment of these HCAb found in Camelidae (Figure 2) are also called VHH to distinguish them from the normal VH that originate from the classical antibodies. When these VHH are recombinantly obtained they are also called “nanobody” because of their size. The difference between VHH and VH originates from a missing CH1, one of the constant domains of the heavy chain, in HCAb [1] (Figure 1b). Besides a heavy chain with a molecular mass that is about 10 to 12 kDa lower this results in a heavy chain lacking the domain required for binding with the light chain. Without this domain the heavy chain binds directly to the hinge region and no light chain is present [10]. Although this CH1 domain is also encoded by the heavy chain gene in HCAb, like in normal antibodies, these HCAb later miss this domain because of a point mutation in the splice site behind the 3’ end of the CH1 exon [10]. Deletion of the CH1 domain for functional HCAb seems to be crucial since the chaperone BiP that retains heavy chains in the endoplasmic reticulum is normal- Figure 2. X-ray structure of a VHH fragment from a HCAb. The part of the scaffold that would normally be interacting with the VL domain in a regular FV is shown in light green. The CDR1, CDR2 and CDR3 domains are shown in red, orange and yellow respectively. Between the CDR3 and CDR1 domain a stick figure is shown to indicate the disulfide bond connecting these two domains. The figure was adapted from Desmyter et al. 1996 [33]. 2 reduced aggregation [13]. Beside these distinct features three other characteristic properties of VHH can be found in their hypervariable complementarity determining regions (CDR). First, VHH have a typically longer CDR3. The domain is on average 17 residues long as opposed to the 12 residues in humans or even 9 in mice [14]. Additionally this CDR3 domain also has an additional disulfide bond, besides the conserved intradomain disulphide bond, that forms a bond with a cysteine in the CDR1 domain [12] (Figure 2). The last feature lies in the hypervariable CDR1 domain that is extended towards the N-terminal end [15] and has an additional hypervariable region at residues 27-30 which are important for somatic mutations and are most likely important for the VHH in binding to antigens [16]. The combination of these longer CDR regions and the occurrence of loop conformations not present in, for instance, human antibodies may provide VHH with a bigger repertoire of antigen binding [15] or perhaps it might help compensate for the lacking VL binding site. pared to conventional antibody derived VH [12], VHH also have a very high stability. A number of VHH that were incubated for one week at 37 °C kept 100% of their activity and others had a still remarkable 80% activity left [17]. VHH overall were found to be very resistant to heat-induced denaturation when they were incubated at temperatures of up to 80 °C over a longer period of time [23]. Some VHH were even capable to still bind antigen specifically at 90 °C. None of the mouse antibodies that where targeting the same antigen where active at this temperature [24]. This higher thermal resistance is most likely due to the decreased hydrophobocity of VHH since ‘humanizing’ VHH to resemble VH resulted in a lower heat resistance [25]. On top of that Dumoulin et al. found that besides thermal resistance, activity could also be fully restored after chemical denaturation by high concentrations of guanidinium chloride and urea [26]. The high robustness against denaturing conditions might in the future prove to be useful for the use of nanobodies outside of therapeutics in industry and research. Also the refolding after denaturation of VHH could prove to be advantageous in large scale protein production when recombinant nanobodies are produced in inclusion bodies and will have to be diluted and refolded before use. One of the key features of VHH, that might be of great use in the future, is the fact that besides their nanomolar affinity (reviewed by [27]) they have the capability of binding unique epitopes. This originates from the adaptations of their hypervariable regions that had to be introduced due to the absence of a light chain and thereby three missing CDRs [12]. The originating distinct loop conformations and longer CDR3 domain allow VHH to create epitope binding surface areas that are as large as normal VH-VL pairs have and bind into grooves and cavities of enzymes (reviewed by [28]) that conventional antibodies because of their flat or slightly grooved surface cannot reach [29]. In fact, VHH even seem to prefer to penetrate these active grooves in enzymes [30]. Therefore VHH or nanobodies could for instance also be used as enzyme inhibitors since these cavities in enzymes can play an important role in their enzymatic activity. This kind of inhibitory activity has already been Advantages of VHH The smallest intact antibody derived fragments, Fv or scFv (Figure 1), that can be generated from the normal antibodies are about 30 kDa. However, recombinant VHH, or nanobodies, can be as small as 15 kDa. Because these nanobodies only consist of a single chain they have a short time from gene to protein when they are recombinantly being expressed. This could be seen by the high levels of protein when recombinant single domain VHH were expressed in Escherichia coli [17] and even higher levels could be seen in Saccharomyces cerevisiae [18]. All of these levels are at least 10 times higher than that what could be obtained with scFv constructs [17]. The later mentioned yeast production might however not be favorable in therapeutics because they cannot modify human glycosylation structures. Furthermore, yeast specific oligosaccharides are added that result in increased immunogenicity (reviewed by [19]). Production has however also been proven to be capable in animal cells [20], plants [21] and insect cells [22]. Besides the mentioned advantage of the increased solubility as a result from certain amino acid conformations com3 proven to work in carbonic anhydrase [31] and lysozyme [32,33]. It was also observed that nanobodies in ELISA experiments blocked substrate cleavage which further proved that these antibodies can have enzyme inhibitory effects [31]. The property of binding to unconventional epitopes in combination with their size will allow nanobodies to penetrate tissue more effectively than conventional antibodies and bind to epitopes that these antibodies would not be able to reach nor bind. It has to be noted however that this small size, although having several advantages, also has its drawbacks. Molecules that are smaller in size than approximately 60 kDa (estimated kidney clearance threshold) have a high renal clearance. A short half-life might however also prove to be useful or even essential when for instance nanobodies are used as targeting mechanism and are coupled to toxic molecules (reviewed by [34]). In this case exposure to healthy cells must be kept to a minimum and a long half-life would be a major drawback. However, an increased half-life is preferred in many other applications. The solution to this problem however is relatively easy; make the nanobody bigger. This has proven to work by fusing nanobodies to long lived proteins like albumin [35] or immunoglobulin [36], another antibody targeting one of these long lived proteins [37] or adding them to a chemical like polyethylene glycol [38]. In all cases this greatly increased the halflife of the nanobodies. Although VHH derived nanobodies seem to be so different from normal VH, because they have so much more suitable properties over conventional antibodies, the overall homology between VH and VHH is quite high. The effective result is that VHH, unlike for instance mouse antibodies that can cause an immune response [39], would be more easily applicable in therapeutics. Although it is possible to combine antibody sequences of humans and mice to reduce the immune response of murine derived antibodies [40], it is easier to “humanize” VHH because of their relatively high similarity to VH. A disadvantage of VHH is however that on their own they cannot elicit functions as antibody-dependent cytotoxicity or complementdependent cytolysis due to their missing Fc domains. Bispecific nanobodies however, who will be discussed later in more detail, offer the possibility of restoring these functions [41]. Obtaining antigen specific nanobodies Several techniques can be applied to obtain antigen specific antibodies. The most common ones are the direct cloning of the VHH genes from B-cells obtained from peripheral blood or lymph nodes [42] and the creation of synthetic libraries. Both of these have their advantages and disadvantages. For the direct cloning of VHH genes first the immunization of a HCAb producing animal like a llama or dromedary is needed (Llamas are often preferred though since they are easier to keep because of their size and are easier to immunize [43]). mRNA then has to be isolated from the blood for cDNA synthesis (Figure 3A-C). This can be amplified by a single set of primers in a PCR (Figure 3D) since all VHH belong to the same family and are encoded by a single exon [15]. These genes differ between the various B-cells but always have the same homologous border sequences. It is important though to select primers that do not amplify the VH genes that also occur in Camelidae. Because of the hydrophobic surface of VH that is exposed with the missing VL fragment, the VH fragments aggregate more easily, resulting in aspecific binding during the selection of specific antibodies [12,13,44]. Another possibility is of course to select the PCR fragments on their size since VHH fragments with their missing CH1 domain (Figure 1b) have shorter fragments. Although there are multiple techniques that can be used, the mostly preferred option to select specific antibodies is the use of a phage display (reviewed by [45]). A phage display library can be constructed by cloning the obtained VHH DNA fragments in a phage genome (Figure 3E) where they are fused to one of the genes that encodes for a coat protein [17]. After transfection (Figure 3E) and assembly of these phages in a bacterium this will result in the VHH being expressed on the coat of the phage. This then allows for panning (binding to an immobilized antigens) (Figure 3F). After a wash step only the phages that express a VHH specific for the immobilized antigen will remain bound (Figure 3F). These wash steps can also be repeated multiple times with different conditions to 4 select for the highest specificity and stability. Since the remaining phages still have the genetic material of the expressed VHH they can then be used to infect bacteria so their DNA coding for the VHH can be used for subsequent steps (reviewed by [46]). This technique of selecting VHH is similar as used for conventional antibodies with the drawback that these consist of 2 variable domains that work together. The VH en VL domain both need to be amplified with different primer sets and cloned into constructs. Furthermore the linking of these 2 fragments in a construct is done randomly since the original combination no library has to be constructed to find the antigen specific antibody-fragment combination. VHH therefore have the advantage in this application since they consist of just one variable domain, reducing the amount of work needed to obtain antigen specific antibodies. In synthetic libraries the first steps of obtaining the cDNA are quite similar (Figure 3A-D). The main difference though is that this can also be done without prior immunization of the animal. Non-immune libraries of VHH have in this way been created [48,49]. With the same panning and selection as previously described antibodies against a desired antigen can be obtained. The downside of this however is that the affinity compared to antibodies derived from an immunized library is low (in the micromolar range [50] while this normally is in the nanomolar range (reviewed by [27])). These affinities can however be improved by in vitro affinity maturation (Figure 3D-2). The antigen binding capabilities of these antibodies are (partially) build artificially in this technique. Random mutations are introduced in the CDR regions of the antigen with for instance an error prone PCR and splicing and shuffling of these sequences [51]. By selection of the desired antigen via panning and then repeating the mutation steps VHH with increased affinity can be obtained in a way that mimics the in vivo affinity maturation. By using different selection procedures also VHH with different physical properties like heat resistance or solubility and stability [50] can be obtained. Synthetic libraries have several advantages over their immunized counterparts. One of these for instance is not having to purify proteins to immunize animals, that are costly to keep and maintain. Not to mention the constant ethical debates about using animals for scientific purposes. More importantly however is that in this way antibodies can be obtained against antigens that could not be used to immunize animals because they are toxic [52] or pathogenic [51]. Furthermore immune libraries are normally made for a specific single antigen whereas synthetic libraries can be applied more readily to a bigger variety of antigens. The downside is however the inherent low affinity of antibodies obtained via non-immunized libraries. Figure 3. Schematic representation of key steps to obtain specific VHH. (A) Blood is obtained from a HCAb producing animal. (B) mRNA is isolated and (C) used for cDNA synthesis. (D) primers specific for VHH are used to amplify VHH coding genes. (D2) When VHH with a higher affinity are to be created an additional step with in vitro affinity purification can be performed before (E) cloning the genes in the phage display vectors and transforming them to bacteria to create a gene library. When using immunized animals this step is usually not needed. (F) The phages created by the bacteria are then panned on immobilized antigen and washed to obtain those with specific binding. (G) The remaining phages are then used to transfect bacteria so the DNA coding for the specific VHH can be used in subsequent steps. longer exists [47]. Therefore a much larger 5 They therefore need (time consuming) affinity maturation steps. er antibodies that can bind these antibodies mimic the original epitope, the catalytic site of the enzyme. These are called catalytic antibodies, or abzymes, and can mimick the enzymatic active site of an enzyme [56]. As a model system nanobodies were used in this way to create abzymes with alliinase activity that could, in combination with alliin, suppress tumor growth in vitro [57]. A perhaps less obvious application, but also worth mentioning function of nanobodies is in aiding the formation of crystals for x-ray crystallography. The main bottleneck in x-ray crystallography is the formation of the crystals. Although natural partner proteins can alleviate this problem by stabilizing these proteins these are not always available. Alternative binders like nanobodies with their unique binding properties offer a solution. Besides stabilizing complexes [58] and organizing disordered proteins [59] for crystal formation, the property of nanobodies to bind to active sites [30] have also allowed the identification of proteins in an active conformation. Nanobodies were able to lock the β2 adrenergic receptor in that state by mimicking an agonistic G protein that are normally absent because of their instability [60]. Other ways in which nanobodies have been helpful is by stabilizing flexible multidomain proteins into more rigid hetero tetramer formations and thereby improving the crystal formation [61]. To illustrate to what extend nanobodies aid the formation of crystals; an EpsI-EpsJ dimer crystal could be made 20 times faster (15 days instead of 10 months) with the help of nanobodies [62]. Uses for nanobodies So far nanobodies or VHH have been described to be advantageous because of their high expression, solubility and stability, high specificity, epitope binding properties unseen by conventional antibodies, immunogenicity and more. Al these additional (and beneficial) properties on top of the ‘normal’ features of conventional antibodies open up a whole array of possible uses for antibodies. Because of these varying features nanobodies have already been suggested for applications in different fields. These range from the possible use as probes for the measurement of caffeine in hot beverages because of their high thermal stability [53] to the use as compounds to increase the antibiotic sensitivity of bacteria [54] because of their enzyme inhibitory functions [28]. These are however just a few of the possible ways that exploit the features of nanobodies. Unique binding properties One of the potential uses of nanobodies is in the use of anti-idiotypic agents [27]. These type of agents function as a replicate of the original antigen (originating from e.g. a tumor associated antigen) and can for instance be used in a vaccine (reviewed by [55]). The distinct structures of the antigen-binding site of VHH might make nanobodies ideal candidates since they could target epitopes that VL-VH combinations could not. More importantly however, for these type of vaccinations it is important that the immune response is targeted to the CDR region and not the scaffold. Here lies the other advantage of nanobodies. While their CDR region is so distinct from conventional antibodies VHH do show a high similarity with VH fragments. This should shift the hosts immune response from the scaffold to the more important CDR region that is mimicking the antigen. Besides their use as antiidiotypic agents in vaccines anti-idiotypic nanobodies can also be used for different purposes. The property of nanobodies to preferentially bind in the active sites of enzymes [30] makes them ideal for the production of catalytic anti-idiotypic antibodies. Oth- Stability Because of their high conformational stability [26] nanobodies are ideal for oral uptake since they have the ability to survive the harsh conditions in the stomach and intestines. Proteolytic stable nanobodies could therefore be selected that were not degraded in vivo and were able to bind the diarrhea causing Escherichia coli in pigs [63]. Similar results were obtained with nanobodies that were able to survive the acidic environment of mice intestine and could still target the diarrhea causing rotavirus [64]. In a completely different example the high stability of nanobodies was found 6 to be of use in shampoo for the treatment of dandruff. Nanobodies were able to survive the surfactants present in the shampoo and retain their ability to bind to Malassezia furfur, a dandruff causing fungus [65]. This kind of stability make nanobodies also ideal candidates as capturing agents in affinity columns [66,67] and biosensors (reviewed by [68]) to name just a few of the various possibilities. er (reviewed by [73]). scFvs are made by linking VH and VL chains together. These linkers, although they can be improved [74], are susceptible to proteolysis and subsequent aggregation. Therefore, fusing two scFvs together, what will increase the number of linkers needed from one to three, is not desirable. Nanobodies however seem to again have the ideal properties to improve on this technique (Figure 4B). The by default monomeric nature of nanobodies reduces the number of linkers needed. The creation of bi-specific nanobodies has therefore already shown to be promising with solubility and expression levels that are comparable to the monomeric variants [75]. In a different experiment the power of bi-specific nanobodies have been shown by viral targeting that was improved up to a 4000-fold after the fusion of nanobodies that targeted different epitopes [76]. Other reasons why nanobodies are preferred for this application are their stability, solubility, ease of production and naturally their size. Bivalent nanobodies are still five times smaller than monomeric intact antibodies, thereby still keeping their better tissue penetrating capabilities. Multivalent antibodies are not only created by fusing two different antibodies together though. Size Besides the discussed properties like their unique binding abilities and high stability the feature that makes nanobodies so supreme in many other applications is their small size. Besides properties as increased tissue penetration, reaching epitopes conventional antibodies cannot [69] and even showing promise of crossing the blood-brain barrier [70], the small size and consequential simple structure make nanobodies ideal building blocks to be fused to other molecules thereby further increasing their capabilities. As described before [35,36] nanobodies can be made into bivalent constructs with the goal of increasing their half-life (Figure 4A). Bivalent constructs with conventional antibodies have however been created before with different goals, namely in the form of bispecific antibodies [71] (Figure 4B). These types of antibodies have numerous possible applications in therapeutics. Multiple epitopes on an antigen could be targeted at the same time, for instance, increasing their affinity and specificity. Other applications could be in the treatment of cancer if bispecific antibodies can first bind immune cells with one of their antibodies and consequently help recruit these cells to tumor cells with the help of their other antibody. This type of format is also called a crosslinking reagent. Bispecific (normal) antibodies with this goal have indeed been tested and reports claimed better results than with the monomeric counterparts [72]. Of course this kind of application could also be used with other types of diseased cells. Where conventional antibodies however already had the disadvantage of being big, and thus having reduced tissues penetrating capabilities, these multivalent versions have this problem doubled. Bi-specific antibodies have therefore been made by fusing the smaller scFvs togeth- Figure 4. Examples of bivalent nanobodies. (A) A VHH fragment and a long lived protein like albumin for example can be fused together to obtain nanobodies with a longer half-life. (B) Other possibilities are the fusion of 2 VHH fragments, either targeting the same or different epitopes, to increase their affinity or recruit cells to a desired location for instance. (C) Finally, a commonly used technique is the fusion of VHH to toxins or drugs like certain enzymes that are needed at specific locations. With the help of VHH these can be targeted there. 7 Fusing nanobodies that target the same epitope together for instance to form dimers or trimers has already been shown to increase their potency up to a 4000-fold when going from a monomer to a trimer by the company Ablynx, the eponym of the nanobody [77]. As of late even pentamers have been created that could increase the binding a 1000 to 10,000 fold for low affinity antibodies and showed promise in the use of antigen discovery [78,79]. Other applications for these multimers can be found in the use of synthetic library derived antibodies [49] who often have a low binding affinity. Coupling of nanobodies to other molecules to increase their half-life or affinity have been mentioned (Figure 4A). There are however more possible uses when it comes to coupling nanobodies to different molecules. Drugs, like those used in chemotherapy, might be very effective in killing their targets. The targeting on itself however will not always be as precise. Antibodies on the other hand can be extremely specific in their targeting, but a single domain antibody like a VHH on its own will then again not have the same results as the drug. The combination of these types of molecules therefore seems a logical next step in the creation of bifunctional constructs (Figure 4C). This approach of combining a selective targeting molecule to a, for that same target, toxic molecule, is also called a “magic bullet”. This approach with immunofusions has been tested before with conventional antibodies. Already in 1988 Senter et al. fused antibodies to alkaline phosphatase, an enzyme able to convert etoposide phosphate into the antitumor drug etoposide, resulting in an antitumor activity that could not have been achieved by administering the drug on its own [80] showing the validity of this type of application. Because these types of antibodies consist of multiple domains that need to be in the right conformation for proper functioning fusion to a toxin is therefore less straightforward than with the monomeric nanobodies. Fusions of nanobodies to toxins have already been successfully tested in varying applications. In an effort to increase the selectivity of the in oral care used antimicrobial agent glucose oxidase this drug was fused to VHH targeting Streptococcus mutans. Consequently S. mutans was found to be more susceptible to this “magic bullet” than the toxin on its own. Although the overall protection was still low the hybrid molecule could selectively target Streptococcus mutans and locally increase antimicrobial activity [81]. Besides bacteria also drug resistant parasites, causing sleeping sickness, have been successfully targeted by this approach. Because of the unique features of nanobodies the cryptic and less immunogenic epitopes of Trypanosoma brucei rhodesiense could be targeted [82]. Fusing these VHH with apolipoprotein L-1, a protein that lyses trypanosome, resulted in clearance of the parasite [83]. These bivalent constructs are however not all based on the fusion of a nanobody to a toxic molecule. Rothbauer et al. showed that nanobodies are also ideal in diagnostic tools when fused to a fluorescent molecule. With a new proof of principle they showed that these nanobodies, when targeting endogenous proteins, allowed for the tracing of the targeted antigens in living cells [84]. Another example of nanobodies fused to a non-toxic compound, and that more importantly shows how diverse their application can be, is seen in the laundry industry where cellulose targeting VHH have been found to more efficiently target fragrances to clothes [85]. This just goes to show that the possible uses for these multivalent formats are enormous. Since the technique behind the fusion of nanobodies to other molecules is more or less the same for all of above mentioned fusions it are the same advantages as in other nanobody based constructs that make them suitable for the use in “magic bullets”. Their monomeric nature for instance makes the fusion of a bivalent construct genetically more straightforward. This is more problematic with regular antibodies because of the multiple domains that are needed in the right conformation for proper functioning. This monomeric nature combined with their small size is also the reason why nanobodies easily fold to their natural conformation when expressed in vivo. Complete antibodies and also scFv fragments on the other hand are already more complex than nanobodies. The disulfide bonds needed between the two scFv fragments for instance do not always form in vivo depending on the in8 tracellular targeting. scFv targeted to the ER for instance have their disulfide bonds formed, but the ones targeted to the cytosol do not [86]. It is therefore that nanobodies might be good candidates to be used as intrabodies, antibodies that are expressed intracellular. Intrabodies are a powerful method to bind and inhibit functions of targeted proteins in vivo (reviewed by [87]). Their potential was first seen in Saccharomyces cerevisiae where cDNA encoding for a light- and heavy-chain against alcohol dehydrogenase I neutralized its enzyme activity in vivo [88]. The possible use for these intrabodies in therapeutics was then also shown by an increased virus infection protection in transgenic plants expressing a scFv antibody without assembly requirements against essential viral proteins [89]. These scFv have later also been used in human cell lines with for instance the inhibition of an influenza A virus [90]. Although nanobodies have not been used as intrabodies as often as scFv have in studies they do already show great promise. In planta for instance they were found to fold properly, were able to target different organelles and most importantly were able to bind to enzymes to inhibit their function [91]. Nanobody derived intrabodies were the first to be used in in vivo studies in mammals and showed proof of principle by inhibiting the secretion of hepatitis B in mice [92]. Nanobodies expressed intracellular have even shown to have potential in the treatment of HIV-1. In infected cells nanobodies could bind to Rev, a protein controlling the expression of HIV proteins, and block the formation of Rev multimers. This prevented the production of viral HIV-1 [93]. As mentioned scFv as intrabodies have already been tested in more varying applications. These range from a possible use in therapeutics against multiple sclerosis, certain types of cancer [94] and the treatment of protein misfolding diseases like prion diseases [95], parkinsons disease (reviewed by [96]) and huntington’s disease [97] to different approaches in the treatment of HIV-1 [98] as opposed to the nanobody variant. The advantages that nanobodies offer over scFv are however very likely to shift future research from scFv to nanobodies as they also open up other strategies for intrabodies in regard to their unique binding epitopes and enzyme inhibitory functions. The only drawback here that has to be overcome, by all intrabodies, is the transfection of the genetic material in animals. Namely, these in vivo transfection efficiencies are still very low. So although intrabodies offer many possible strategies in treating a variety of diseases the main problem mentioned is not with the nanobodies, but in getting to the required location. Getting these nanobodies intracellular might however be a problem that can be solved with nanobodies themselves. This will be discussed in more detail in the following chapter. (Possible) applications for nanobodies in cancer treatment Conventional antibodies have already been used in the treatment of different diseases for several years. As mentioned before however these antibodies have several downsides, or lacking features, compared to nanobodies. The usage of nanobodies as new therapeutic antibodies is therefore expected to greatly increase upon the efficiency and possible new applications compared to conventional antibodies. However, the treatment of diseases with antibodies requires the targeting of specific markers. Cancer for instance will be a disease subjected to many drug trials with nanobodies. This is reflected by the fact that already over 50% of the conventional antibodies that are in development are meant for cancer treatment (reviewed by [99]). But what is a cancer marker, and is there even such a thing? Some characteristics for a tumor marker would obviously be that they are consistently (over) expressed on tumor cells and ideally not on healthy cells (reviewed by [100]). A number of these type of markers that allow for the distinguishing between cancer and healthy cells are already in use by current monoclonal antibodies (reviewed by [101]). Nanobodies have however also proven to be excellent at tumor targeting with properties like high specificity, rapid clearance of excess antibodies and the fact that they are nonimmunogenic [102]. Some of the previously mentioned markers have therefore also been targeted by nanobodies. For instance, nanobodies have been tested that target the carcinoembryonic antigen (CEA) [103], epidermal 9 growth factor receptor (EGFR) [104], human epidermal growth factor receptor-2s (HER2) [105] and mucine 1 (MUC1) [106] to name just a few. With the advent of nanobodies however a whole new array of markers might be exploited that could previously not be targeted. The unique features that nanobodies offer will likely improve upon existing therapies and possibly open up whole new possibilities in the treatment of cancer. Here the advantages of nanobodies and the possibilities that these open for varying applications are discussed in regard to current and future applications for cancer treatment. The most important thing before antibodies can target their specific antigens is of course getting to that antigen. Here the advantage of the nanobodies small size comes into play. Tissue penetration is an obstacle were many conventional antibodies have to deal with. And although 85% of all cancers are solid, only a relative small percentage of cancer drugs based on antibodies are targeting these solid tumors. This is probably due to the increased difficulty of targeting these types of tissue (reviewed by [107]). The higher resistance to these drugs originates from the extracellular matrix of tumors that is more difficult to pass through because of extracellular matrix proteins [108]. Also the slower blood flow in tumors complicates their targeting [109]. Therefore to effectively penetrate tumor tissue the size of molecules is of great importance. This can be seen by the diffusion rate between complete IgG molecules and scFv fragments. The later can diffuse 12 times faster than whole IgG antibodies (6 hours vs 30 min) [110]. This higher penetration rate is however paid for by a shorter retention, also due to their size. As mentioned before however, this does not have to be a bad thing. Their small size opens up to a plethora of other beneficial properties besides higher tumor penetration that can be used in the treatment of cancer. One of the advantages of having a good tumor penetration and fast clearance for instance is of use in tumor markers. These are necessary for the first step in the treatment of cancer, namely a rapid and accurate diagnosis. For this application a long retention is not necessary and the properties of nanobodies have already shown promise in the diagnosis of prostate cancer [42] and breast cancer [111] for instance. Monomeric nature of nanobodies The EGFR is a tumor associated antigen (TAA) already targeted by many different antibodies in several studies [112,113]. Nanobodies against EGFR have now also been studied [104] because they are easy to identify and select for in phage display libraries and offer many advantages for follow up experiments like fusion to different types of molecules because of their monovalent nature. The nanobodies selected in these experiments were found to block EGF mediated signaling and thereby inhibit cell proliferation. Besides offering a new targeting mechanism for EGFR this study shows that the monomeric nature of nanobodies allow for the fast development of antigen specific antibodies [104]. These properties of nanobodies can also proof to be of use in perhaps less obvious applications. It has been shown for instance that certain bacteria like bifidobacterium, clostridium, and salmonella preferably target tumors over healthy tissue to replicate in [114]. This feature could be further exploited by the use of nanobodies. Since it is already known that nanobodies can be readily expressed in vivo in different expression systems [17,18,2022] because of their (simple) monomeric nature and hydrophilicity, expression in these type of bacteria might also be possible. And since it is also known that intact nanobodies can be transported across the outer membrane [115] these nanobodies could be presented on the cell wall to help in the targeting of tumor cells. After insertion of the bacteria in the tumor other nanobodies that are expressed and secreted from these bacteria could aid in killing tumor cells by targeting certain cancer markers. This might prove to be beneficial since the nanobodies targeting these tumor markers are expressed in the location where they are needed. This might potentially not only increase their efficacy but also reduce possible side effects. A similar technique with cells that target tumors and express nanobodies has already been tested recently. This was done with stem cells instead of bacteria. These stem cells could target brain tumors and secrete EGFR targeting 10 nanobodies on the desired location resulting in a significant reduction of tumor growth [116]. The proposed targeting of cells by expressing antibodies on their surface is also a technique that, on its own, has already proven to work. Cytotoxic T cells have been targeted to tumors with antibodies that are presented extracellular. These antibodies, linked to an extracellular spacer domain and an intracellular signaling domain, are able to activate the T cell upon binding (reviewed by [117]). This has normally been done with the use of scFv, the use of nanobodies might however be preferred. And indeed T cells with nanobodies instead of scFv to help with their targeting have already been successfully directed to tumor cells by MUC1 targeting [118]. All in all directing cells with the help of nanobodies and the secretion of functional nanobodies are strategies that have been proven to work separately. When combined in one cell, as proposed with the bacteria, they might however have synergistic effects that could benefit both techniques. As mentioned the use of intrabodies shows great potential in inhibition of targeted proteins. Getting there however might prove to be more problematic. Nanobodies could be able to help with both of these points. Bispecific scFv have been found to be able to target DNA to tumor cells in the form of adenoviral vectors [119]. As mentioned though, scFv bivalent constructs have some disadvantages that bivalent nanobodies do not have because of their monomeric nature. Therefore by using nanobodies in these constructs instead of scFv the targeting on itself could be improved. Another method wherein nanobodies can help in the targeting of genes to cancer cells is with non-viral vectors. Such non-viral vectors consisting of polyethylenimine (PEI) and polyethylene glycol (PEG) for example are very efficient at transfecting cells but are not very specific in their targeting. Nanobodies however have been found very useful for this application when covalently linked to the PEG. MUC1 targeting nanobodies in combination with a PEG-PEI conjugate were in this way able to transfect a plasmid coding for a killer gene behind the cancer specific MUC1 promoter [120]. This resulted in cell death in tumor cell lines. Without the MUC1 promoter this did not happen indicating that the genetic code was indeed transfected and the non-viral transfection with PEG and PEI itself was not the reason for cell death [120]. Besides transfecting killer genes this could obviously also be used for the transfection of a gene expressing an intrabody against an intracellular cancer marker to increase the specificity. One of these intracellular targets could for instance be the transactivation domain of the c-Myc gene, a gene that is constitutively expressed in many cancers and regulates cell proliferation among other things (reviewed by [121]). This domain is already known to be targetable by scFv antibodies [122]. These scFv were however not internally expressed but translocated across the membrane via a fusion to a protein transduction domain resulting in a cell-permeable antibody. This strategy however required very high amounts of fused antibodies [122]. Intracellular expression with nanobodies might improve upon this. Namely, the fact that intracellular markers can indeed be targeted by intracellular expressed nanobodies has been shown by the targeting of the (possible) metastasis marker hnRNP-K (heterogeneous nuclear ribonucleoprotein K) [123]. Bivalent nanobodies The monovalent nature of nanobodies does not only make them suitable for the rapid search of new antibodies against certain targets, but as mentioned they can also be made bivalent more easily than for instance scFv. Bivalent conventional antibodies have already been proven to be advantageous in the treatment of cancer [72]. But also homo dimeric nanobodies [104] as well as hetero dimeric nanobodies targeting adjacent epitopes [124] have shown to improve binding to EGFR compared to their monovalent versions. Other types of hetero dimeric nanobodies, or bispecific nanobodies, that target completely different epitopes have been found to be able to bind NK-cells and activate interferongamma production. Together with the right tumor marker these bi-specific nanobodies show promise of being able to recruit NK-cells to kill tumor cells [125]. The ease with which these bivalent constructs can be made could greatly improve upon the targeting of tumor 11 cells. The production of bivalent nanobodies will namely be more appealing since the resulting constructs are more soluble and stable and therefore will show more promise to eventually end up as a future cancer therapy. and inhibit enzymes [30]. This property can be of use in cancer therapy since some enzymes play key roles in tumor growth like ribonucleotide reductase (reviewed by [128]) and topoisomerase II (reviewed by [129]) which has already been targeted with antibodies [80]. Although some of these enzymes can also be targeted by drugs, the use of nanobodies might offer more possibilities in terms of delivery and fusion constructs. For enzymes that have not yet been able to be targeted by drugs the use of nanobodies is more obvious because of the relative ease to create and find new nanobodies that can target a desired antigen. Since some of these enzymes are however located on the inside of cells this strategy might have to be combined with gene therapy like the before mentioned example with the PEG-PEI conjugate. Instead of the in that example used killer gene a different gene coding for an intracellular cancer marker could be used. This could further increase the therapy’s specificity since nanobodies first need to select for extracellular cancer markers before the intracellular targeting nanobodies can do their job. Bifunctional nanobodies As described, nanobodies can be fused to toxic molecules relatively easy compared to their conventional counterparts. This might be especially useful in cancer treatment since nanobodies in this way can help increase the specificity of chemotherapy thereby increasing its efficacy and reducing the side effects by keeping the exposure to healthy tissue to a minimum. Also because of the fact that they have a fast blood clearance nanobodies have been fused to radio nuclides that emit radiation and result in the destruction of cells. Nanobodies targeting the human epidermal growth-factor receptor type 2 have in this way been fused to the radio-metal 177Lu without affecting the antigen recognition of the nanobody and having a higher tumor uptake compared to healthy tissue [126]. Similar results have been obtained by fusing 177Lu to EGFR targeting nanobodies. Additionally these nanobodies were also fused to albumin what resulted in a greatly reduced blood clearance, but it also vastly improved the tumor uptake [127]. This goes to show that a consideration has to be made between fast clearance or better uptake, depending on the type of application. Other successes in the treatment of tumor cells with bifunctional nanobodies have been gained by the fusion of β-lactamase to carcinoembryonic antigen targeting VHH. The selective binding of the VHH to tumor cells resulted in a locally increased toxic drug concentration resulting from the β-lactamase that converted a nontoxic prodrug to its toxic variant [103]. The authors also reported that a rapid clearance of the multivalent construct from non-targeted tissue was seen, without the need of clearance agents. This again shows the advantage of the nanobodies small size. Opinion and perspectives Since the discovery of HCAb in camels almost 20 years ago VHH fragments or nanobodies have been used in a growing number of applications. Mainly their small size and monomeric nature make them superior in many ways compared to conventional antibodies. Furthermore nanobodies are very stable, easily expressed and functional in vivo, ideal to create multivalent constructs and can recognize unique epitopes that the conventional antibodies cannot. And these are just a few of their advantages. The so far only possible downside is the potential immunogenicity that might be elicited when nanobodies are fused into larger constructs. On their own they seem to cause no significant immunological reaction but larger constructs are known to cause a stronger response. This is something that will have to be tested in the future since the fusion of nanobodies to various molecules is one of the features that make them so desirable for many applications. Most likely this will be no problem though since the fusion of these molecules to conventional antibodies would Unique binding properties Some of the properties like the longer CDR3 domain give nanobodies the unique binding properties that allow them to more easily bind 12 result in even larger constructs making nanobodies still the better option. The low cost of producing nanobodies opens up possible uses in consumer products like the already mentioned shampoo and detergent. Their stability allows oral uptake, making them suitable for instance to be mixed in feed for animals and their unique binding properties will allow easier use of antiidiotopic agents, for example. It is however their simple monomeric nature and size that will show the true capabilities of nanobodies. This might especially hold true in cancer diagnosis and therapeutics. As mentioned the largest part of all cancers consists of solid tumors. The difficult targeting of these tumors however causes a comparatively low number of drugs to be designed for this purpose. Nanobodies have shown to be more capable in penetrating these tumors. With their antigen binding properties and/or drug delivery possibilities they will be able to target a wider variety of (solid) tumors. One of the techniques that can be used in cancer therapeutics and shows a lot of potential with the use of nanobodies are intrabodies. The only thing standing in the way of a real breakthrough in therapeutics for this type of application is getting these antibodies inside the cell. Gene therapy would be the most ideal solution but still has downsides like low efficiency. But even in the targeting of genes to specific cells nanobodies have shown great potential for the future. This just goes to show the wide variety of applications nanobodies can be used in. Their broad spectra of applications is probably also the reason for the constantly growing interest in these type of antibodies. Conventional antibodies have been in use for a longer period of time, explaining why they are still more often found in various applications. However, the many different uses wherein nanobodies have already proven to be superior to conventional antibodies and the potential they offer in other applications make it likely that many future therapeutics and applications will be based on nanobodies rather than "normal" antibodies. The prediction is therefore that nanobodies will eventually be preferred in the development of new applications, thereby gradually expanding the uses for antibodies and likely replacing the conventional antibodies. References 1. Padlan EA: Anatomy of the antibody molecule. Molecular Immunology 1994, 31:169-217. 2. Inbar D, Hochman J, Givol D: Localization of antibody-combining sites within the variable portions of heavy and light chains. Proc Natl Acad Sci U S A 1972, 69:2659-2662. 3. Huston JS, Levinson D, Mudgett-Hunter M, Tai MS, Novotny J, Margolies MN, Ridge RJ, Bruccoleri RE, Haber E, Crea R, et al.: Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. Proc Natl Acad Sci U S A 1988, 85:5879-5883. 4. Ward ES, Gussow D, Griffiths AD, Jones PT, Winter G: Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli. Nature 1989, 341:544546. 5. Yoo TJ, Roholt OA, Pressman D: Specific binding activity of isolated light chains of antibodies. Science 1967, 157:707-709. 6. Borrebaeck CAK, Malmborg A-C, Furebring C, Michaelsson A, Ward S, Danielsson L, Ohlin M: Kinetic Analysis of Recombinant Antibody-Antigen Interactions: Relation Between Structural Domains and Antigen Binding. Nat Biotech 1992, 10:697-698. 7. Alexander A, Steinmetz M, Barritault D, Frangione B, Franklin EC, Hood L, Buxbaum JN: gamma Heavy chain disease in man: cDNA sequence supports partial gene deletion model. Proc Natl Acad Sci U S A 1982, 79:3260-3264. 8. Greenberg AS, Avila D, Hughes M, Hughes A, McKinney EC, Flajnik MF: A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature 1995, 374:168-173. 9. Hamers-Casterman C, Atarhouch T, Muyldermans S, Robinson G, Hamers C, Songa EB, Bendahman N, Hamers R: Naturally occurring antibodies devoid of light chains. Nature 1993, 363:446-448. 10. Khong Nguyen V, Hamers R, Wyns L, Muyldermans S: Loss of splice consensus signal is responsible for the removal of the entire CH1 domain of the functional 13 camel IGG2A heavy-chain antibodies. Molecular Immunology 1999, 36:515-524. 11. Lee YK, Brewer JW, Hellman R, Hendershot LM: BiP and immunoglobulin light chain cooperate to control the folding of heavy chain and ensure the fidelity of immunoglobulin assembly. Mol Biol Cell 1999, 10:2209-2219. 12. Muyldermans S, Atarhouch T, Saldanha J, Barbosa JARG, Hamers R: Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. Protein Engineering 1994, 7:1129-1135. 13. Davies J, Riechmann L: ‘Camelising’ human antibody fragments: NMR studies on VH domains. FEBS Letters 1994, 339:285-290. 14. Wu TT, Johnson G, Kabat EA: Length distribution of CDRH3 in antibodies. Proteins: Structure, Function, and Bioinformatics 1993, 16:1-7. 15. Vu KB, Ghahroudi MA, Wyns L, Muyldermans S: Comparison of llama VH sequences from conventional and heavy chain antibodies. Molecular Immunology 1997, 34:11211131. 16. Nguyen VK, Hamers R, Wyns L, Muyldermans S: Camel heavy-chain antibodies: diverse germline V(H)H and specific mechanisms enlarge the antigen-binding repertoire. EMBO J 2000, 19:921-930. 17. Arbabi Ghahroudi M, Desmyter A, Wyns L, Hamers R, Muyldermans S: Selection and identification of single domain antibody fragments from camel heavy-chain antibodies. FEBS Letters 1997, 414:521526. 18. Frenken LGJ, van der Linden RHJ, Hermans PWJJ, Bos JW, Ruuls RC, de Geus B, Verrips CT: Isolation of antigen specific Llama VHH antibody fragments and their high level secretion by Saccharomyces cerevisiae. Journal of Biotechnology 2000, 78:11-21. 19. Gerngross TU: Advances in the production of human therapeutic proteins in yeasts and filamentous fungi. Nat Biotech 2004, 22:1409-1414. 20. Zhang J, Liu X, Bell A, To R, Baral TN, Azizi A, Li J, Cass B, Durocher Y: Transient expression and purification of chimeric heavy chain antibodies. Protein Expression and Purification 2009, 65:77-82. 21. Ismaili A, Jalali-javaran M, Rasaee MJ, Rahbarizadeh F, Forouzandeh-moghadam M, Memari HR: Production and characterization of anti-(mucin MUC1) single-domain antibody in tobacco (Nicotiana tabacum cultivar Xanthi). Biotechnology and Applied Biochemistry 2007, 047:11-19. 22. Gomez-Sebastian S, Nunez MC, Garaicoechea L, Alvarado C, Mozgovoj M, Lasa R, Kahl A, Wigdorovitz A, Parreno V, Escribano JM: Rotavirus A-specific single-domain antibodies produced in baculovirusinfected insect larvae are protective in vivo. BMC Biotechnol 2012, 12:59. 23. Ewert S, Cambillau C, Conrath K, Plückthun A: Biophysical Properties of Camelid VHH Domains Compared to Those of Human VH3 Domains†. Biochemistry 2002, 41:3628-3636. 24. van der Linden RHJ, Frenken LGJ, de Geus B, Harmsen MM, Ruuls RC, Stok W, de Ron L, Wilson S, Davis P, Verrips CT: Comparison of physical chemical properties of llama VHH antibody fragments and mouse monoclonal antibodies. Biochimica et Biophysica Acta (BBA) - Protein Structure and Molecular Enzymology 1999, 1431:3746. 25. Conrath K, Vincke C, Stijlemans B, Schymkowitz J, Decanniere K, Wyns L, Muyldermans S, Loris R: Antigen binding and solubility effects upon the veneering of a camel VHH in framework-2 to mimic a VH. J Mol Biol 2005, 350:112-125. 26. Dumoulin M, Conrath K, Van Meirhaeghe A, Meersman F, Heremans K, Frenken LGJ, Muyldermans S, Wyns L, Matagne A: Single-domain antibody fragments with high conformational stability. Protein Science 2002, 11:500-515. 27. Muyldermans S, Lauwereys M: Unique singledomain antigen binding fragments derived from naturally occurring camel heavy-chain antibodies. Journal of Molecular Recognition 1999, 12:131-140. 28. Muyldermans S, Cambillau C, Wyns L: Recognition of antigens by single-domain antibody fragments: the superfluous luxury of paired domains. Trends in Biochemical Sciences 2001, 26:230-235. 29. Padlan EA: X-Ray Crystallography of Antibodies. In Advances in Protein Chemistry. Edited by Frederic M. Richards DEE, Peter SK: Academic Press; 1996:57133. vol Volume 49.] 30. De Genst E, Silence K, Decanniere K, Conrath K, Loris R, Kinne J, Muyldermans S, Wyns L: Molecular basis for the preferential cleft recognition by dromedary heavy-chain 14 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. antibodies. Proc Natl Acad Sci U S A 2006, 103:4586-4591. Lauwereys M, Arbabi Ghahroudi M, Desmyter A, Kinne J, Holzer W, De Genst E, Wyns L, Muyldermans S: Potent enzyme inhibitors derived from dromedary heavy-chain antibodies. EMBO J 1998, 17:3512-3520. Transue TR, De Genst E, Ghahroudi MA, Wyns L, Muyldermans S: Camel single-domain antibody inhibits enzyme by mimicking carbohydrate substrate. Proteins 1998, 32:515-522. Desmyter A, Transue TR, Ghahroudi MA, Thi MH, Poortmans F, Hamers R, Muyldermans S, Wyns L: Crystal structure of a camel single-domain VH antibody fragment in complex with lysozyme. Nat Struct Biol 1996, 3:803-811. Carter P: Improving the efficacy of antibodybased cancer therapies. Nat Rev Cancer 2001, 1:118-129. Vosjan MJ, Vercammen J, Kolkman JA, Stigtervan Walsum M, Revets H, van Dongen GA: Nanobodies targeting the hepatocyte growth factor: potential new drugs for molecular cancer therapy. Mol Cancer Ther 2012, 11:1017-1025. Harmsen MM, Van Solt CB, Fijten HP, Van Setten MC: Prolonged in vivo residence times of llama single-domain antibody fragments in pigs by binding to porcine immunoglobulins. Vaccine 2005, 23:49264934. Dennis MS, Zhang M, Meng YG, Kadkhodayan M, Kirchhofer D, Combs D, Damico LA: Albumin Binding as a General Strategy for Improving the Pharmacokinetics of Proteins. Journal of Biological Chemistry 2002, 277:35035-35043. Harmsen MM, van Solt CB, Fijten HP, van Keulen L, Rosalia RA, Weerdmeester K, Cornelissen AH, De Bruin MG, Eble PL, Dekker A: Passive immunization of guinea pigs with llama single-domain antibody fragments against foot-and-mouth disease. Vet Microbiol 2007, 120:193-206. Courtenay-Luck NS, Epenetos AA, Moore R, Larche M, Pectasides D, Dhokia B, Ritter MA: Development of primary and secondary immune responses to mouse monoclonal antibodies used in the diagnosis and therapy of malignant neoplasms. Cancer Res 1986, 46:64896493. Boulianne GL, Hozumi N, Shulman MJ: Production of functional chimaeric mouse/human antibody. Nature 1984, 312:643-646. 41. Holliger P, Wing M, Pound JD, Bohlen H, Winter G: Retargeting serum immunoglobulin with bispecific diabodies. Nat Biotechnol 1997, 15:632-636. 42. Saerens D, Kinne J, Bosmans E, Wernery U, Muyldermans S, Conrath K: Single Domain Antibodies Derived from Dromedary Lymph Node and Peripheral Blood Lymphocytes Sensing Conformational Variants of Prostate-specific Antigen. Journal of Biological Chemistry 2004, 279:51965-51972. 43. van der Linden R, de Geus B, Stok W, Bos W, van Wassenaar D, Verrips T, Frenken L: Induction of immune responses and molecular cloning of the heavy chain antibody repertoire of Lama glama. Journal of Immunological Methods 2000, 240:185-195. 44. Davies J, Riechmann L: Antibody VH Domains as Small Recognition Units. Nat Biotech 1995, 13:475-479. 45. Hoogenboom HR: Selecting and screening recombinant antibody libraries. Nat Biotech 2005, 23:1105-1116. 46. Hoogenboom HR, de Bruı ̈ne AP, Hufton SE, Hoet RM, Arends J-W, Roovers RC: Antibody phage display technology and its applications. Immunotechnology 1998, 4:1-20. 47. Winter G, Griffiths AD, Hawkins RE, Hoogenboom HR: Making Antibodies by Phage Display Technology. Annual Review of Immunology 1994, 12:433-455. 48. Tanha J, Dubuc G, Hirama T, Narang SA, MacKenzie CR: Selection by phage display of llama conventional VH fragments with heavy chain antibody VHH properties. Journal of Immunological Methods 2002, 263:97-109. 49. Verheesen P, Roussis A, de Haard HJ, Groot AJ, Stam JC, den Dunnen JT, Frants RR, Verkleij AJ, Theo Verrips C, van der Maarel SM: Reliable and controllable antibody fragment selections from Camelid nonimmune libraries for target validation. Biochimica et Biophysica Acta (BBA) Proteins and Proteomics 2006, 1764:13071319. 50. Yau KYF, Dubuc G, Li S, Hirama T, MacKenzie CR, Jermutus L, Hall JC, Tanha J: Affinity maturation of a VHH by mutational hotspot randomization. Journal of Immunological Methods 2005, 297:213224. 15 51. Goldman ER, Anderson GP, Liu JL, Delehanty JB, Sherwood LJ, Osborn LE, Cummins LB, Hayhurst A: Facile generation of heatstable antiviral and antitoxin single domain antibodies from a semisynthetic llama library. Anal Chem 2006, 78:82458255. 52. Goldman ER, Anderson GP, Conway J, Sherwood LJ, Fech M, Vo B, Liu JL, Hayhurst A: Thermostable llama single domain antibodies for detection of botulinum A neurotoxin complex. Anal Chem 2008, 80:8583-8591. 53. Ladenson RC, Crimmins DL, Landt Y, Ladenson JH: Isolation and characterization of a thermally stable recombinant anticaffeine heavy-chain antibody fragment. Anal Chem 2006, 78:4501-4508. 54. Conrath KE, Lauwereys M, Galleni M, Matagne A, Frere JM, Kinne J, Wyns L, Muyldermans S: Beta-lactamase inhibitors derived from single-domain antibody fragments elicited in the camelidae. Antimicrob Agents Chemother 2001, 45:2807-2812. 55. Bhattacharya-Chatterjee M, Chatterjee SK, Foon KA: Anti-idiotype antibody vaccine therapy for cancer. Expert Opin Biol Ther 2002, 2:869-881. 56. Friboulet A, Izadyar L, Avalle B, Roseto A, Thomas D: Abzyme generation using an anti-idiotypic antibody as the “internal image” of an enzyme active site. Applied Biochemistry and Biotechnology 1994, 47:229-239. 57. Li J-W, Xia L, Su Y, Liu H, Xia X, Lu Q, Yang C, Reheman K: Molecular Imprint of Enzyme Active Site by Camel Nanobodies: RAPID AND EFFICIENT APPROACH TO PRODUCE ABZYMES WITH ALLIINASE ACTIVITY. Journal of Biological Chemistry 2012, 287:13713-13721. 58. Rasmussen SGF, DeVree BT, Zou Y, Kruse AC, Chung KY, Kobilka TS, Thian FS, Chae PS, Pardon E, Calinski D, et al.: Crystal structure of the [bgr]2 adrenergic receptor-Gs protein complex. Nature 2011, 477:549-555. 59. Loris R, Marianovsky I, Lah J, Laeremans T, Engelberg-Kulka H, Glaser G, Muyldermans S, Wyns L: Crystal Structure of the Intrinsically Flexible Addiction Antidote MazE. Journal of Biological Chemistry 2003, 278:28252-28257. 60. Rasmussen SGF, Choi H-J, Fung JJ, Pardon E, Casarosa P, Chae PS, DeVree BT, Rosenbaum DM, Thian FS, Kobilka TS, et al.: Structure of a nanobody-stabilized active state of the [bgr]2 adrenoceptor. Nature 2011, 469:175-180. 61. Korotkov KV, Pardon E, Steyaert J, Hol WG: Crystal structure of the N-terminal domain of the secretin GspD from ETEC determined with the assistance of a nanobody. Structure 2009, 17:255-265. 62. Lam AY, Pardon E, Korotkov KV, Hol WGJ, Steyaert J: Nanobody-aided structure determination of the EpsI:EpsJ pseudopilin heterodimer from Vibrio vulnificus. Journal of Structural Biology 2009, 166:8-15. 63. Harmsen MM, Solt CB, Zijderveld-van Bemmel AM, Niewold TA, Zijderveld FG: Selection and optimization of proteolytically stable llama single-domain antibody fragments for oral immunotherapy. Applied Microbiology and Biotechnology 2006, 72:544-551. 64. van der Vaart JM, Pant N, Wolvers D, Bezemer S, Hermans PW, Bellamy K, Sarker SA, van der Logt CPE, Svensson L, Verrips CT, et al.: Reduction in morbidity of rotavirus induced diarrhoea in mice by yeast produced monovalent llama-derived antibody fragments. Vaccine 2006, 24:4130-4137. 65. Dolk E, van der Vaart M, Lutje Hulsik D, Vriend G, de Haard H, Spinelli S, Cambillau C, Frenken L, Verrips T: Isolation of llama antibody fragments for prevention of dandruff by phage display in shampoo. Appl Environ Microbiol 2005, 71:442-450. 66. Klooster R, Maassen BTH, Stam JC, Hermans PW, ten Haaft MR, Detmers FJM, de Haard HJ, Post JA, Theo Verrips C: Improved anti-IgG and HSA affinity ligands: Clinical application of VHH antibody technology. Journal of Immunological Methods 2007, 324:1-12. 67. Verheesen P, ten Haaft MR, Lindner N, Verrips CT, de Haard JJW: Beneficial properties of single-domain antibody fragments for application in immunoaffinity purification and immuno-perfusion chromatography. Biochimica et Biophysica Acta (BBA) - General Subjects 2003, 1624:21-28. 68. Saerens D, Huang L, Bonroy K, Muyldermans S: Antibody Fragments as Probe in Biosensor Development. Sensors 2008, 8:4669-4686. 69. Strokappe N, Szynol A, Aasa-Chapman M, Gorlani A, Forsman Quigley A, Hulsik DL, Chen L, Weiss R, de Haard H, Verrips T: 16 Llama Antibody Fragments Recognizing Various Epitopes of the CD4bs Neutralize a Broad Range of HIV-1 Subtypes A, B and C. PLoS One 2012, 7:e33298. 70. Muruganandam A, Tanha J, Narang S, Stanimirovic D: Selection of phagedisplayed llama single-domain antibodies that transmigrate across human bloodbrain barrier endothelium. The FASEB Journal 2001. 71. Milstein C, Cuello AC: Hybrid hybridomas and their use in immunohistochemistry. Nature 1983, 305:537-540. 72. Segal DM, Weiner GJ, Weiner LM: Bispecific antibodies in cancer therapy. Current Opinion in Immunology 1999, 11:558-562. 73. Hudson PJ: Recombinant antibody constructs in cancer therapy. Current Opinion in Immunology 1999, 11:548-557. 74. Whitlow M, Bell BA, Feng SL, Filpula D, Hardman KD, Hubert SL, Rollence ML, Wood JF, Schott ME, Milenic DE, et al.: An improved linker for single-chain Fv with reduced aggregation and enhanced proteolytic stability. Protein Eng 1993, 6:989-995. 75. Els Conrath K, Lauwereys M, Wyns L, Muyldermans S: Camel single-domain antibodies as modular building units in bispecific and bivalent antibody constructs. J Biol Chem 2001, 276:73467350. 76. Hultberg A, Temperton NJ, Rosseels V, Koenders M, Gonzalez-Pajuelo M, Schepens B, Ibanez LI, Vanlandschoot P, Schillemans J, Saunders M, et al.: Llamaderived single domain antibodies to build multivalent, superpotent and broadened neutralizing anti-viral molecules. PLoS One 2011, 6:e17665. 77. Kolkman JA, Law DA: Nanobodies – from llamas to therapeutic proteins. Drug Discovery Today: Technologies 2010, 7:e139-e146. 78. Zhang J, Tanha J, Hirama T, Khieu NH, To R, Tong-Sevinc H, Stone E, Brisson J-R, Roger MacKenzie C: Pentamerization of Singledomain Antibodies from Phage Libraries: A Novel Strategy for the Rapid Generation of High-avidity Antibody Reagents. J Mol Biol 2004, 335:49-56. 79. Zhang J, Li Q, Nguyen T-D, Tremblay T-L, Stone E, To R, Kelly J, Roger MacKenzie C: A Pentavalent Single-domain Antibody Approach to Tumor Antigen Discovery and the Development of Novel Proteomics Reagents. J Mol Biol 2004, 341:161-169. 80. Senter PD, Saulnier MG, Schreiber GJ, Hirschberg DL, Brown JP, Hellstrom I, Hellstrom KE: Anti-tumor effects of antibody-alkaline phosphatase conjugates in combination with etoposide phosphate. Proc Natl Acad Sci U S A 1988, 85:4842-4846. 81. Szynol A, de Soet JJ, Sieben-van Tuyl E, Bos JW, Frenken LG: Bactericidal effects of a fusion protein of llama heavy-chain antibodies coupled to glucose oxidase on oral bacteria. Antimicrob Agents Chemother 2004, 48:3390-3395. 82. Stijlemans B, Conrath K, Cortez-Retamozo V, Van Xong H, Wyns L, Senter P, Revets H, De Baetselier P, Muyldermans S, Magez S: Efficient targeting of conserved cryptic epitopes of infectious agents by single domain antibodies. African trypanosomes as paradigm. J Biol Chem 2004, 279:1256-1261. 83. Baral TN, Magez S, Stijlemans B, Conrath K, Vanhollebeke B, Pays E, Muyldermans S, De Baetselier P: Experimental therapy of African trypanosomiasis with a nanobody-conjugated human trypanolytic factor. Nat Med 2006, 12:580-584. 84. Rothbauer U, Zolghadr K, Tillib S, Nowak D, Schermelleh L, Gahl A, Backmann N, Conrath K, Muyldermans S, Cardoso MC, et al.: Targeting and tracing antigens in live cells with fluorescent nanobodies. Nat Meth 2006, 3:887-889. 85. Lewis W, Keshavarz-Moore E, Windust J, Bushell D, Parry N: Construction and evaluation of novel fusion proteins for targeted delivery of micro particles to cellulose surfaces. Biotechnology and Bioengineering 2006, 94:625-632. 86. Biocca S, Ruberti F, Tafani M, Pierandrel-Amaldi P, Cattaneo A: Redox State of Single Chain Fv Fragments Targeted to the Endoplasmic Reticulum, Cytosol and Mitochondria. Nat Biotech 1995, 13:11101115. 87. Richardson JH, Marasco WA: Intracellular antibodies: development and therapeutic potential. Trends in Biotechnology 1995, 13:306-310. 88. Carlson JR: A new means of inducibly inactivating a cellular protein. Mol Cell Biol 1988, 8:2638-2646. 89. Tavladoraki P, Benvenuto E, Trinca S, De Martinis D, Cattaneo A, Galeffi P: 17 Transgenic plants expressing a functional single-chain Fv antibody are specifically protected from virus attack. Nature 1993, 366:469-472. 90. Mukhtar MM, Li S, Li W, Wan T, Mu Y, Wei W, Kang L, Rasool ST, Xiao Y, Zhu Y, et al.: Single-chain intracellular antibodies inhibit influenza virus replication by disrupting interaction of proteins involved in viral replication and transcription. The International Journal of Biochemistry & Cell Biology 2009, 41:554560. 91. Jobling SA, Jarman C, Teh M-M, Holmberg N, Blake C, Verhoeyen ME: Immunomodulation of enzyme function in plants by single-domain antibody fragments. Nat Biotech 2003, 21:77-80. 92. Serruys B, Van Houtte F, Verbrugghe P, LerouxRoels G, Vanlandschoot P: Llama-derived single-domain intrabodies inhibit secretion of hepatitis B virions in mice. Hepatology 2009, 49:39-49. 93. Vercruysse T, Pardon E, Vanstreels E, Steyaert J, Daelemans D: An intrabody based on a llama single-domain antibody targeting the N-terminal alpha-helical multimerization domain of HIV-1 rev prevents viral production. J Biol Chem 2010, 285:21768-21780. 94. Strebe N, Guse A, Schüngel M, Schirrmann T, Hafner M, Jostock T, Hust M, Müller W, Dübel S: Functional knockdown of VCAM1 at the posttranslational level with ER retained antibodies. Journal of Immunological Methods 2009, 341:30-40. 95. Cardinale A, Filesi I, Vetrugno V, Pocchiari M, Sy M-S, Biocca S: Trapping Prion Protein in the Endoplasmic Reticulum Impairs PrPC Maturation and Prevents PrPSc Accumulation. Journal of Biological Chemistry 2005, 280:685-694. 96. Zhou C, Przedborski S: Intrabody and Parkinson's disease. Biochim Biophys Acta 2009, 1792:634-642. 97. Colby DW, Chu Y, Cassady JP, Duennwald M, Zazulak H, Webster JM, Messer A, Lindquist S, Ingram VM, Wittrup KD: Potent inhibition of huntingtin aggregation and cytotoxicity by a disulfide bond-free single-domain intracellular antibody. Proc Natl Acad Sci U S A 2004, 101:17616-17621. 98. Bai J, Sui J, Zhu RY, Tallarico ASC, Gennari F, Zhang D, Marasco WA: Inhibition of Tatmediated Transactivation and HIV-1 Replication by Human Anti-hCyclinT1 Intrabodies. Journal of Biological Chemistry 2003, 278:1433-1442. 99. Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC: Monoclonal antibody successes in the clinic. Nat Biotech 2005, 23:10731078. 100. Carter P, Smith L, Ryan M: Identification and validation of cell surface antigens for antibody targeting in oncology. Endocrine-Related Cancer 2004, 11:659687. 101. Reichert JM, Valge-Archer VE: Development trends for monoclonal antibody cancer therapeutics. Nat Rev Drug Discov 2007, 6:349-356. 102. Cortez-Retamozo V, Lauwereys M, Hassanzadeh Gh G, Gobert M, Conrath K, Muyldermans S, De Baetselier P, Revets H: Efficient tumor targeting by singledomain antibody fragments of camels. International Journal of Cancer 2002, 98:456-462. 103. Cortez-Retamozo V, Backmann N, Senter PD, Wernery U, De Baetselier P, Muyldermans S, Revets H: Efficient Cancer Therapy with a Nanobody-Based Conjugate. Cancer Res 2004, 64:2853-2857. 104. Roovers R, Laeremans T, Huang L, Taeye S, Verkleij A, Revets H, Haard H, Bergen en Henegouwen PMP: Efficient inhibition of EGFR signalling and of tumour growth by antagonistic anti-EGFR Nanobodies. Cancer Immunology, Immunotherapy 2007, 56:303-317. 105. Sheikholeslami F, Rasaee MJ, Shokrgozar MA, Dizaji MM, Rahbarizadeh F, Ahmadvande D: Isolation of a Novel Nanobody Against HER-2/neu Using Phage Displays Technology. Lab Medicine 2010, 41:69-76. 106. Rahbarizadeh F, Rasaee MJ, Forouzandeh Moghadam M, Allameh AA, Sadroddiny E: Production of novel recombinant singledomain antibodies against tandem repeat region of MUC1 mucin. Hybrid Hybridomics 2004, 23:151-159. 107. Jang S, Wientjes MG, Lu D, Au JS: Drug Delivery and Transport to Solid Tumors. Pharmaceutical Research 2003, 20:13371350. 108. Jain RK: Transport of Molecules in the Tumor Interstitium: A Review. Cancer Res 1987, 47:3039-3051. 109. Jain RK: Physiological Barriers to Delivery of Monoclonal Antibodies and Other Macromolecules in Tumors. Cancer Res 1990, 50:814s-819s. 18 110. Graff CP, Wittrup KD: Theoretical Analysis of Antibody Targeting of Tumor Spheroids: Importance of Dosage for Penetration, and Affinity for Retention. Cancer Res 2003, 63:1288-1296. 111. Vaneycken I, Devoogdt N, Van Gassen N, Vincke C, Xavier C, Wernery U, Muyldermans S, Lahoutte T, Caveliers V: Preclinical screening of anti-HER2 nanobodies for molecular imaging of breast cancer. FASEB J 2011, 25:24332446. 112. Herbst RS, Arquette M, Shin DM, Dicke K, Vokes EE, Azarnia N, Hong WK, Kies MS: Phase II Multicenter Study of the Epidermal Growth Factor Receptor Antibody Cetuximab and Cisplatin for Recurrent and Refractory Squamous Cell Carcinoma of the Head and Neck. Journal of Clinical Oncology 2005, 23:5578-5587. 113. Foon KA, Yang X-D, Weiner LM, Belldegrun AS, Figlin RA, Crawford J, Rowinsky EK, Dutcher JP, Vogelzang NJ, Gollub J, et al.: Preclinical and clinical evaluations of ABX-EGF, a fully human anti-epidermal growth factor receptor antibody. International journal of radiation oncology, biology, physics 2004, 58:984990. 114. Pawelek JM, Low KB, Bermudes D: Bacteria as tumour-targeting vectors. The Lancet Oncology 2003, 4:548-556. 115. Veiga E, De Lorenzo V, Fernández LA: Structural tolerance of bacterial autotransporters for folded passenger protein domains. Molecular Microbiology 2004, 52:1069-1080. 116. van de Water JAJM, Bagci-Onder T, Agarwal AS, Wakimoto H, Roovers RC, Zhu Y, Kasmieh R, Bhere D, Van Bergen en Henegouwen PMP, Shah K: Therapeutic stem cells expressing variants of EGFRspecific nanobodies have antitumor effects. Proceedings of the National Academy of Sciences 2012. 117. Eshhar Z: The T-Body Approach: Redirecting T Cells with Antibody Specificity. In Therapeutic Antibodies. Edited by Chernajovsky Y, Nissim A: Springer Berlin Heidelberg; 2008:329-342. Handbook of Experimental Pharmacology, vol 181.] 118. Bakhtiari SH, Rahbarizadeh F, Hasannia S, Ahmadvand D, Iri-Sofla FJ, Rasaee MJ: Anti-MUC1 nanobody can redirect T-body cytotoxic effector function. Hybridoma (Larchmt) 2009, 28:85-92. 119. Haisma HJ, Grill J, Curiel DT, Hoogeland S, van Beusechem VW, Pinedo HM, Gerritsen WR: Targeting of adenoviral vectors through a bispecific single-chain antibody. Cancer Gene Ther 2000, 7:901904. 120. Sadeqzadeh E, Rahbarizadeh F, Ahmadvand D, Rasaee MJ, Parhamifar L, Moghimi SM: Combined MUC1-specific nanobodytagged PEG-polyethylenimine polyplex targeting and transcriptional targeting of tBid transgene for directed killing of MUC1 over-expressing tumour cells. Journal of Controlled Release 2011, 156:85-91. 121. Dang CV: c-Myc Target Genes Involved in Cell Growth, Apoptosis, and Metabolism. Mol Cell Biol 1999, 19:1-11. 122. Avignolo C, Bagnasco L, Biasotti B, Melchiori A, Tomati V, Bauer I, Salis A, Chiossone L, Mingari MC, Orecchia P, et al.: Internalization via Antennapedia protein transduction domain of an scFv antibody toward c-Myc protein. The FASEB Journal 2008, 22:1237-1245. 123. Inoue A, Sawata SY, Taira K, Wadhwa R: Lossof-function screening by randomized intracellular antibodies: identification of hnRNP-K as a potential target for metastasis. Proc Natl Acad Sci U S A 2007, 104:8983-8988. 124. Roovers RC, Vosjan MJ, Laeremans T, el Khoulati R, de Bruin RC, Ferguson KM, Verkleij AJ, van Dongen GA, van Bergen en Henegouwen PM: A biparatopic antiEGFR nanobody efficiently inhibits solid tumour growth. Int J Cancer 2011, 129:2013-2024. 125. Behar G, Sibéril S, Groulet A, Chames P, Pugnière M, Boix C, Sautès-Fridman C, Teillaud J-L, Baty D: Isolation and characterization of anti-FcγRIII (CD16) llama single-domain antibodies that activate natural killer cells. Protein Engineering Design and Selection 2008, 21:1-10. 126. D'Huyvetter M, Aerts A, Xavier C, Vaneycken I, Devoogdt N, Gijs M, Impens N, Baatout S, Ponsard B, Muyldermans S, et al.: Development of 177Lu-nanobodies for radioimmunotherapy of HER2-positive breast cancer: evaluation of different bifunctional chelators. Contrast Media & Molecular Imaging 2012, 7:254-264. 127. Tijink BM, Laeremans T, Budde M, Walsum MS-v, Dreier T, de Haard HJ, Leemans CR, van Dongen GAMS: Improved tumor 19 targeting of anti–epidermal growth factor receptor Nanobodies through albumin binding: taking advantage of modular Nanobody technology. Mol Cancer Ther 2008, 7:2288-2297. 128. Wijerathna SR, Ahmad MF, Xu H, Fairman JW, Zhang A, Kaushal PS, Wan Q, Kiser J, Dealwis CG: Targeting the Large Subunit 129. 20 of Human Ribonucleotide Reductase for Cancer Chemotherapy. Pharmaceuticals 2011, 4:1328-1354. Baldwin EL, Osheroff N: Etoposide, topoisomerase II and cancer. Curr Med Chem Anticancer Agents 2005, 5:363-372.